Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where David Carlile is active.

Publication


Featured researches published by David Carlile.


Blood | 2012

Phase 1 study results of the type II glycoengineered humanized anti-CD20 monoclonal antibody obinutuzumab (GA101) in B-cell lymphoma patients

Gilles Salles; Franck Morschhauser; Thierry Lamy; Noel Milpied; Catherine Thieblemont; Hervé Tilly; Gabriele Bieska; Elina Asikanius; David Carlile; Joe Birkett; Pavel Pisa; Guillaume Cartron

Whereas the chimeric type I anti-CD20 Ab rituximab has improved outcomes for patients with B-cell malignancies significantly, many patients with non-Hodgkin lymphoma (NHL) remain incurable. Obinutuzumab (GA101) is a glycoengineered, humanized anti-CD20 type II Ab that has demonstrated superior activity against type I Abs in vitro and in preclinical studies. In the present study, we evaluated the safety, efficacy, and pharmacokinetics of GA101 in a phase 1 study of 21 patients with heavily pretreated, relapsed, or refractory CD20(+) indolent NHL. Patients received GA101 in a dose-escalating fashion (3 per cohort, range 50/100-1200/2000 mg) for 8 × 21-day cycles. The majority of adverse events (AEs) were grades 1 and 2 (114 of 132 total AEs). Seven patients reported a total of 18 grade 3 or 4 AEs. Infusion-related reactions were the most common AE, with most occurring during the first infusion and resolving with appropriate management. Three patients experienced grade 3 or 4 drug-related infusion-related reactions. The best overall response was 43%, with 5 complete responses and 4 partial responses. Data from this study suggest that GA101 was well tolerated and demonstrated encouraging activity in patients with previously treated NHL up to doses of 2000 mg. This trial is registered at www.clinicaltrials.gov as NCT00517530.


Blood | 2012

A phase 1 study of obinutuzumab induction followed by 2 years of maintenance in patients with relapsed CD20-positive B-cell malignancies

Laurie H. Sehn; Sarit Assouline; Douglas A. Stewart; Joy Mangel; Randy D. Gascoyne; Gregg Fine; Susan Frances-Lasserre; David Carlile; Michael Crump

This phase 1 study evaluated the safety, tolerability, pharmacokinetics, and antitumor activity of obinutuzumab (GA101), a glycoengineered type II anti-CD20 monoclonal antibody administered as induction followed by 2 years of maintenance. Cohorts of 3 to 6 patients received obinutuzumab (200-2000 mg) intravenously weekly for 4 weeks. Patients with a complete or partial response (or stable disease and clinical benefit) continued to receive obinutuzumab every 3 months, for a maximum of 8 doses. Twenty-two patients with relapsed CD20-positive non-Hodgkin lymphoma or chronic lymphocytic leukemia with an indication for treatment and no therapy of higher priority were enrolled. Patients received a median of 4 prior regimens; 86% had received at least 1 rituximab-containing regimen. No dose-limiting or unexpected AEs were observed. Infusion-related reactions were most common (all grades, 73%; grade 3/4, 18%), followed by infection (32%), pyrexia (23%), neutropenia (23%), headache (18%), and nausea (18%). At end of induction, 5 (23%) patients achieved partial responses and 12 (54%) had stable disease. Eight patients received maintenance; best overall response was 32% (6 partial responses/1 complete response). Obinutuzumab induction and maintenance therapy was well tolerated with promising efficacy in this heterogeneous, highly pretreated population and warrants further investigation. This study was registered at www.clinicaltrials.gov (identifier NCT00576758).


Blood | 2016

Randomized phase 2 study of obinutuzumab monotherapy in symptomatic, previously untreated chronic lymphocytic leukemia

John C. Byrd; Joseph M. Flynn; Thomas J. Kipps; Michael Boxer; Kathryn S. Kolibaba; David Carlile; Guenter Fingerle-Rowson; Nicola Tyson; Jamie Hirata; Jeff Porter Sharman

Obinutuzumab is a glycoengineered, type 2 anti-CD20 humanized antibody with single-agent activity in relapsed chronic lymphocytic leukemia (CLL). With other CD20 antibodies, a dose-response relationship has been shown. We therefore performed a randomized phase 2 study in symptomatic, untreated CLL patients to evaluate if an obinutuzumab dose response exists. Obinutuzumab was given at a dose of 1000 mg (100 mg IV day 1, 900 mg day 2, 1000 mg day 8 and day 15 of cycle 1; 1000 mg day 1 of cycles 2-8) or 2000 mg (100 mg IV day 1, 900 mg day 2, 1000 mg day 3, 2000 mg day 8 and day 15 of cycle 1; 2000 mg day 1 of cycles 2-8). The primary end point was overall response rate (ORR). Eighty patients were enrolled with similar demographics: median age 67 years, 41% high-risk Rai disease, and 10% del(17p)(13.1). ORR (67% vs 49%, P = .08) and complete response (CR) or CR with incomplete cytopenia response (20% vs 5%) favored 2000 mg obinutuzumab. Overall, therapy was well tolerated, and infusion events were manageable. This study demonstrates significant efficacy of obinutuzumab monotherapy, for 1000 mg as well as for 2000 mg, in untreated CLL patients with acceptable toxicity. Although exploratory, a dose-response relationship may exist, but its relevance to improving progression-free survival is uncertain and will require further follow-up. This trial was registered at www.clinicaltrials.gov as #NCT01414205.


Haematologica | 2016

Rationale for optimal obinutuzumab/GA101 dosing regimen in B-cell non-Hodgkin lymphoma

Guillaume Cartron; Florence Hourcade-Potelleret; Franck Morschhauser; Gilles Salles; Michael K. Wenger; Anna Truppel-Hartmann; David Carlile

Obinutuzumab (GA101) is a type II, glycoengineered anti-CD20 monoclonal antibody for the treatment of hematologic malignancies. Obinutuzumab has mechanisms of action that are distinct from those of rituximab, potentially translating into improved clinical efficacy. We present the pharmacokinetic and clinical data from the phase I/II GAUGUIN and phase I GAUDI studies that were used to identify the obinutuzumab dose and regimen undergoing phase III assessment. In phase I (GAUGUIN and GAUDI), non-Hodgkin lymphoma patients received up to a maximum 9 fixed doses (obinutuzumab 50–2000 mg). In GAUGUIN phase II, patients received obinutuzumab 400/400 mg or 1600/800 mg [first dose day (D)1, D8, cycle (C) 1; second dose D1, C2–C8]. The influence of demographic factors on pharmacokinetics and drug exposure on tumor response and toxicity were analyzed using exploratory graphical analyses. Obinutuzumab serum concentrations with 1600/800 mg were compared with a 1000 mg fixed-dose regimen (D1, D8 and D15, C1; D1, C2–C8) using pharmacokinetic modeling simulations. Factors related to CD20-antigenic mass were more influential on obinutuzumab pharmacokinetics with 400/400 versus 1600/800 mg. Higher serum concentrations were observed with 1600/800 versus 400/400 mg, irrespective of CD20-antigenic mass. Tumor shrinkage was greater with 1600/800 versus 400/400 mg; there was no significant increase in adverse events. Fixed dose 1000 mg with an additional C1 infusion resulted in similar serum concentrations to 1600/800 mg in model-based analyses. The obinutuzumab 1000 mg fixed-dose regimen identified in this exploratory analysis was confirmed in a full covariate analysis of a larger dataset, and is undergoing phase III evaluation. GAUGUIN and GAUDI are registered at www.clinicaltrials.gov (clinicaltrials.gov identifier:00517530 and 00825149, respectively).


Journal of Cardiovascular Pharmacology | 2012

Evidence of a drug-drug interaction linked to inhibition of ester hydrolysis by orlistat.

Darren Bentley; Anne-Marie Young; Lucy Rowell; Günter Gross; Joseph Tardio; David Carlile

Abstract: Orlistat, a lipase inhibitor taken with meals at doses of 60 mg (available over-the-counter) or 120 mg (prescription only) for treatment of obesity, is known to impair the absorption of fat-soluble molecules. Dalcetrapib, a modulator of cholesteryl ester transfer protein activity, is a lipophilic thioester prodrug. Lipase-induced and pancreatin-induced hydrolysis of dalcetrapib in biorelevant media in vitro was very efficiently inhibited by orlistat. Thus, the potential for orlistat to affect the bioavailability of concomitantly administered dalcetrapib was studied in an open-label 2-cohort study in 24 healthy volunteers as follows: single 600-mg doses of dalcetrapib were administered with increasing doses of orlistat (cohort A: 10, 40, 120 mg; cohort B: 20, 60, 120 mg). Exposure to the active form of dalcetrapib was more than 50% lower when taken with orlistat 60 mg or 120 mg than when taken alone. Similar trends were observed with lower orlistat doses (20 mg and 40 mg). Concomitant administration of orlistat also reduced the pharmacodynamic effects of dalcetrapib treatment on cholesteryl ester transfer protein activity. The interaction exceeds that predicted on the basis of dalcetrapib lipophilicity. These findings demonstrate the potential for large interactions between orlistat and esters that undergo de-esterification in the gastrointestinal tract, independent of lipophilicity.


CPT: Pharmacometrics & Systems Pharmacology | 2015

Challenges and opportunities for quantitative clinical pharmacology in cancer immunotherapy: Something old, something new, something borrowed and something blue

Mark Stroh; David Carlile; Chi-Chung Li; Jonathan Wagg; Benjamin Ribba; Saroja Ramanujan; Jin Jin; Jian Xu; Jean‐Eric Charoin; Zhi‐Xin Xhu; Peter N. Morcos; John D. Davis; Alex Phipps

Cancer immunotherapy (CIT) initiates or enhances the host immune response against cancer. Following decades of development, patients with previously few therapeutic options may now benefit from CIT. Although the quantitative clinical pharmacology (qCP) of previous classes of anticancer drugs has matured during this time, application to CIT may not be straightforward since CIT acts via the immune system. Here we discuss where qCP approaches might best borrow or start anew for CIT.


Journal of drug assessment | 2014

Interaction potential of Carmegliptin with P-glycoprotein (Pgp) transporter in healthy volunteers

Olaf Kuhlmann; David Carlile; Johannes Noe; Darren Bentley

Abstract Objective: The primary objective of this study was to investigate the interaction potential of carmegliptin with P-glycoprotein transporter in vitro and in vivo. A secondary objective was to investigate the safety and tolerability of carmegliptin alone or co-administered with verapamil. Research design and methods: The inhibition potential of carmegliptin was tested in vitro and in a non-randomized open-label study in 16 healthy male volunteers. On day 1 a single dose of carmegliptin (150 mg) was given, followed by a single dose of verapamil (80 mg) on day 7, on day 10 a single dose of carmegliptin (150 mg) together with verapamil (80 mg t.i.d.), and verapamil (80 mg t.i.d.) on days 11–14. Finally, on day 15 a single dose of 150 mg carmegliptin together with 80 mg t.i.d. verapamil was administered. Pharmacokinetic and safety parameters were assessed. Results: Carmegliptin showed in vitro a low cell permeability and was a good substrate for human MDR1 cells. When carmegliptin was taken with verapamil, the mean exposure and Cmax to carmegliptin increased by 29% and 53%, respectively. Increases in exposure were slightly greater on the sixth day of verapamil dosing than on the first day. Verapamil Cmax was 17% lower on average when given with carmegliptin than when verapamil was taken alone, and similar trends were apparent in corresponding norverapamil pharmacokinetics. All reported adverse events (n = 28) were mild in intensity, and verapamil had no apparent effect on the pattern or incidence of events. Conclusions: In vitro, carmegliptin is a substrate but not an inhibitor of human Pgp. Consistently, the co-administration of carmegliptin with verapamil altered the pharmacokinetics of carmegliptin slightly and moderately increased the exposure. Peak exposure of verapamil and its metabolite norverapamil tended to be lower when co-administered with carmegliptin. The combination of carmegliptin and verapamil was generally well tolerated. Although the observed overall changes in pharmacokinetics were small and dose adjustments in clinics are currently not expected, co-administration of carmegliptin with Pgp inhibitors should be carefully monitored in future clinical trials.


Cancer Research | 2012

Abstract LB-220: Translational research with RG7160 (GA201) leads to a phase II clinical study in combination with FOLFIRI in 2nd line metastatic colorectal cancer (mCRC)

A. Cervantes; Ben Markman; Salvatore Siena; Carles Pericay; Giuseppe Aprile; John Bridgewater; Antonio Cubillo; Ashita Waterston; L. Arés; Jean-Charles Soria; S. Temam; David Oppenheim; Jon Chick; David Carlile; Sophie Golding; Maria Longauer Banholzer; Alexandre Passioukov; Chrstian Gerdes; Sophia Soehrman; Christoph Mancao; Luigi Manenti; Josep Tabernero

GA201 is a novel dual-acting humanized, engineered IgG1 anti-EGFR mAb designed to enhance ADCC in combination with signaling inhibition. Superior efficacy was demonstrated versus cetuximab in orthotopic CRC xenograft models. Preclinical data indicated an increase in macrophages (4-5 fold) and NK cells (2-3 fold) infiltration in tumors treated with GA201 compared to cetuximab. In a phase I clinical study objective responses and long lasting disease stabilizations were observed. A marked reduction in circulating NK cells and an increased infiltration of immune cells into skin rash was seen. Preliminary evidence of the enhanced ADCC capacity of GA201 was investigated in 25 third line patients with KRAS-mutant mCRC. Best overall response was SD in 40% of patients and median OS was 9.4 months. Reduction in peripheral NK cells and regulatory T-cells was observed. Comparison of pre- and post-treatment tumour biopsies revealed that tumour-infiltrating immune cells, in particular CD68+ and CD3+ cells increased overall. EGFR-membrane staining in baseline tumour biopsies was markedly higher than in the corresponding archival tumour specimens (median H-score 52 vs 3, respectively), which might reflect technical, but most importantly, biological variability. Based on these results and the commitment to investigate the tumor immune infiltration profile as a potential predictive biomarker, a fresh tumor biopsy at baseline was mandated in the ongoing randomized phase II trial (GAIN-C). This compares GA201 plus irinotecan, infusional fluorouracil and leucovorin (FOLFIRI) with FOLFIRI alone in KRAS-mutant 2nd line mCRC patients and with cetuximab plus FOLFIRI in KRAS-wild type patients (n=160). Primary objective is PFS. The fresh tumor biopsy will be centrally analyzed for EGFR and KRAS. A comprehensive biomarker program was implemented to investigate potential predictive biomarkers, with an emphasis on the real time tumor immune-infiltration status. The safety run-in phase with 36 patients was completed. Most common treatment related AEs on GA201 arms vs cetuximab vs. chemotherapy included (≥ grade 3): rash (33 vs. 17 vs. 0%), IRR (10 vs. 0 vs. 0%), diarrhea (17 vs. 0 vs. 11%), fatigue/asthenia (0 vs. 17 vs. 0%), hypomagnesaemia (17 vs. 0 vs. 0%), stomatitis (17 vs. 0 vs. 0%) and vomiting/nausea (6 vs. 0 vs. 0%). So far, no patient discontinued due to EGFR-related skin toxicity. Immunological profiling is ongoing. Extensive research efforts, including a fresh tumor biopsy in second line mCRC patients, are taken to understand the immunological mechanism of action of GA201 and to select and test (ph. II) and validate (ph. III) potential predictive biomarkers. To guide personalized cancer immunotherapies there is a clear need for reliable biomarkers which would necessitate a tumor assessment directly prior to the therapy and immune monitoring throughout treatment. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr LB-220. doi:1538-7445.AM2012-LB-220


Blood | 2009

Phase I Study of RO5072759 (GA101) in Patients with Relapsed/Refractory CD20+ Non-Hodgkin Lymphoma (NHL).

Gilles Salles; Franck Morschhauser; Thierry Lamy; Noel Milpied; Catherine Thieblemont; Hervé Tilly; Gabi Bieska; David Carlile; Guillaume Cartron


Clinical Pharmacokinectics | 2013

Effect of Hepatic and Renal Impairment on the Pharmacokinetics of Dalcetrapib

Mary Phelan; Judith Anzures-Cabrera; David Carlile; Lucy Rowell; Olaf Kuhlmann; Gerhard Arold; Richard A. Robson; Darren Bentley

Collaboration


Dive into the David Carlile's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge